1
|
Richard Alexander H. Jeffrey Norton and the Legacy of the Surgical Metabolism Section of the Surgery Branch of the NCI. Ann Surg Oncol 2024; 31:3599-3603. [PMID: 38413508 DOI: 10.1245/s10434-024-15078-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 02/08/2024] [Indexed: 02/29/2024]
Affiliation(s)
- H Richard Alexander
- Division of Surgical Oncology, Department of Surgery, Rutgers Robert Wood Johnson Medical School, The Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA.
| |
Collapse
|
2
|
Lyons NB, Proctor KG. Are the Outcomes of a Pig Endotoxemia Model Applicable to Human Sepsis? Crit Care Med 2023; 51:1102-1104. [PMID: 37439647 DOI: 10.1097/ccm.0000000000005904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Affiliation(s)
- Nicole B Lyons
- Both authors: Division of Trauma, Burns, and Surgical Critical Care, Daughtry Family, Department of Surgery, University of Miami Miller School of Medicine, Miami, FL
| | | |
Collapse
|
3
|
Protection against Lipopolysaccharide-Induced Endotoxemia by Terrein Is Mediated by Blocking Interleukin-1β and Interleukin-6 Production. Pharmaceuticals (Basel) 2022; 15:ph15111429. [PMID: 36422559 PMCID: PMC9693353 DOI: 10.3390/ph15111429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/17/2022] [Accepted: 11/17/2022] [Indexed: 11/22/2022] Open
Abstract
Terrein is a fungal metabolite and has been known to exert anti-melanogenesis, anti-cancer, and anti-bacterial activities. However, its role in endotoxemia has never been investigated until now. In the present study, we examined the effect of terrein on lipopolysaccharide (LPS)-induced endotoxemia in mice and characterized the potential mechanisms of action. Treatment with terrein increased the survival of mice and decreased the production of inflammatory cytokines, including interleukin-1β (IL-1β) and interleukin-6 (IL-6) in an LPS-induced endotoxemia model. In addition, terrein suppressed the LPS-induced production of IL-1β and IL-6 in RAW 264.7 cells, a murine macrophage-like cell line, and the mRNA expression of IL-1β and IL-6 was also inhibited by terrein in LPS-stimulated RAW 264.7 cells. Further study demonstrated that terrein blocked LPS-induced phosphorylation of p65 subunit of nuclear factor (NF)/κB and the phosphorylation of c-Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase (MAPK) was also suppressed by terrein treatment. Collectively, these results suggest that terrein exerts a protective effect again LPS-induced endotoxemia in mice by blocking the production of inflammatory cytokines. Our results also suggest that the anti-inflammatory effect of terrein might be mediated, at least in part, by blocking the activation of NF-κB, JNK, and p38 MAPK signaling pathways.
Collapse
|
4
|
Zhang Z, Cao P, Fang M, Zou T, Han J, Duan Y, Xu H, Yang X, Li QS. Design, synthesis, and SAR study of novel 4,5-dihydropyrazole-Thiazole derivatives with anti-inflammatory activities for the treatment of sepsis. Eur J Med Chem 2021; 225:113743. [PMID: 34403978 DOI: 10.1016/j.ejmech.2021.113743] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 08/01/2021] [Accepted: 08/02/2021] [Indexed: 12/24/2022]
Abstract
Systemic inflammatory response syndrome is a major feature of sepsis which is one of the major causes of death worldwide. It has been reported that 3,5-diaryl-4,5-dihydropyrazole and thiazole derivatives have many biological functions, especially in the aspect of anti-inflammation. According to the strategy of pharmacophore combination, we introduced thiazole moiety into dihydropyrazole skeleton to design and synthesize a novel series of 2-(3,5-diphenyl-4,5-dihydro-1H-pyrazol-1-yl)-4-methylthiazole derivatives, and evaluated their anti-inflammatory activities for sepsis treatment. Preliminary structure-activity relationship (SAR) analysis was conducted by their inhibitory activities against nitric oxide (NO) release in LPS-induced RAW264.7 cells, and the optimal compound E26 exhibited more potent anti-inflammatory activity than the positive control treatment indomethacin and dexamethasone. In further mechanism study, our results showed that compound E26 significantly suppressed the production of interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-α), NO and inhibited the expressions of inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2) through blocking MAPKs signaling pathway. In addition, in vivo administration of compound E26 resulted in a significant improvement of LPS-induced sepsis in C57BL/6J mice, with reducing toxicity in multiple organs. Taken together, this study demonstrated the compound E26 could be a promising agent for the treatment of sepsis.
Collapse
Affiliation(s)
- Zhen Zhang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Peichang Cao
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Mengyuan Fang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Tingfeng Zou
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Jihong Han
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Yajun Duan
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Huajian Xu
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Xiaoxiao Yang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China.
| | - Qing-Shan Li
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China.
| |
Collapse
|
5
|
Bahrambeigi S, Khatamnezhad M, Asri-Rezaei S, Dalir-Naghadeh B, Javadi S, Mirzakhani N. Pro-oxidant and degenerative effects of haloperidol under inflammatory conditions in rat; the involvement of SIRT1 and NF-κB signaling pathways. VETERINARY RESEARCH FORUM : AN INTERNATIONAL QUARTERLY JOURNAL 2021; 12:175-183. [PMID: 34345383 PMCID: PMC8328260 DOI: 10.30466/vrf.2019.105811.2514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 07/09/2019] [Indexed: 06/13/2023]
Abstract
The present study was conducted to evaluate the effects of different doses of haloperidol (HP) on induction of oxidative stress in blood and liver cell degeneration in comparison with influences of HP pre-treatment on inflammatory process induced by intraperitoneal (IP) administration of lipopolysaccharide (LPS). One hundred twenty male albino Wistar rats were randomly divided into eight groups (15 in each), including: Control group, LPS group, three groups as HP administration in three divided doses (0.50, 1.00 and 2.00 mg kg-1), and three treatment groups that HP was administered in three doses (0.50, 1.00 and 2.00 mg kg-1) prior to LPS administration. Concentrations of malondialdehyde, activities of antioxidant enzymes including glutathione peroxidase, superoxide dismutase and also the levels of tumor necrosis factor-alpha and interleukin 1-beta were measured in blood and serum. In addition to liver histopathological changes evaluation, hepatic silent information regulator of transcription 1 (SIRT1) and phosphorylated-nuclear factor-κB (p-NF-κB) levels were quantitated. Our findings indicated that sole administration of HP (particularly higher doses) can induce oxidative stress in blood and cell degeneration in liver, while it can attenuate inflammatory process induced by LPS administration presumably via SIRT1 up-regulation and preventing the induction of p-NF-κB. The oxidative and degenerative effects of HP and its impact on inflammatory status were completely dose- dependent according to our results. The possible anti-inflammatory effects of HP may affect reparative mechanisms and hepatic cell degeneration. However, the influences of HP on immune system need further investigations and its higher doses should be administered cautiously especially in patients with immune system dysfunctions.
Collapse
Affiliation(s)
- Saman Bahrambeigi
- Department of Internal Medicine and Clinical Pathology, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran
| | - Mahsa Khatamnezhad
- Department of Internal Medicine and Clinical Pathology, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran
| | - Siamak Asri-Rezaei
- Department of Internal Medicine and Clinical Pathology, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran
| | - Bahram Dalir-Naghadeh
- Department of Internal Medicine and Clinical Pathology, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran
| | - Shahram Javadi
- Department of Internal Medicine and Clinical Pathology, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran
| | - Navideh Mirzakhani
- Department of Pathobiology, Faculty of Veterinary Medicine, Amol University of Special Modern Technologies, Amol, Iran
| |
Collapse
|
6
|
Lee SY, Hsin LW, Su MJ, ChangChien CC, Ku HC. A novel isoquinoline derivative exhibits anti-inflammatory properties and improves the outcomes of endotoxemia. Pharmacol Rep 2019; 71:1281-1288. [PMID: 31683199 DOI: 10.1016/j.pharep.2019.06.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 05/27/2019] [Accepted: 06/28/2019] [Indexed: 12/25/2022]
Abstract
BACKGROUND Sepsis initiates an inflammatory response that causes widespread injury, and candidates for related myocardial depressant factors include cytokines and nitric oxide (NO). Nuclear factor kappa-B (NF-κB) stimulated by toll-like receptor 4 activation in sepsis mediates the transcription of multiple proinflammatory genes. These inflammatory mediators can cause myocardial dysfunction, which may deteriorate sepsis outcomes. To address this risk, we investigated the potential beneficial effects of a novel isoquinolines derivative, CYY054c, in LPS-induced inflammatory response leading to endotoxemia. METHODS The effects of CYY054c on cytokine and inflammatory-related protein production were evaluated in lipopolysaccharide (LPS)-stimulated macrophages. To determine whether CYY054c alleviates inflammatory storm-induced myocardial dysfunction in vivo, LPS was injected in rats, and cardiac function was measured by a pressure-volume loop. RESULTS CYY054c inhibited LPS-induced NF-κB expression in macrophages and reduced the release of tumor necrosis factor-alpha (TNF-α), interleukin-1β (IL-1β), and interleukin-6 (IL-6), as well as the expression of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2). In the animal studies, CYY054c alleviated LPS-upregulated plasma TNF-α, IL-1β, IL-6, and NO concentrations, as well as cardiac monocyte chemotactic protein-1, iNOS, and COX-2 expression in rats, contributing to the improvement of cardiac function during endotoxemia. CONCLUSIONS The reduction of NF-κB-mediated inflammatory mediators and the maintenance of hemodynamic performance by CYY054c improved the outcomes during endotoxemia. CYY054c may be a potential therapeutic agent for sepsis.
Collapse
Affiliation(s)
- Shih-Yi Lee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Taitung MacKay Memorial Hospital, Taiwan; Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, MacKay Memorial Hospital, Taiwan; Mackay Junior College of Medicine, Nursing, and Management, Taipei, Taiwan
| | - Ling-Wei Hsin
- School of Pharmacy, Molecular Probes Development Core, Molecular Imaging Center, Taipei, Taiwan; Center for Innovative Therapeutics Discovery, National Taiwan University, Taipei, Taiwan
| | - Ming-Jai Su
- Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ching-Chia ChangChien
- Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hui-Chun Ku
- Department of Life Science, Fu Jen Catholic University, New Taipei City, Taiwan.
| |
Collapse
|
7
|
Zingarelli B, Coopersmith CM, Drechsler S, Efron P, Marshall JC, Moldawer L, Wiersinga WJ, Xiao X, Osuchowski MF, Thiemermann C. Part I: Minimum Quality Threshold in Preclinical Sepsis Studies (MQTiPSS) for Study Design and Humane Modeling Endpoints. Shock 2019; 51:10-22. [PMID: 30106874 PMCID: PMC6296871 DOI: 10.1097/shk.0000000000001243] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Preclinical animal studies are mandatory before new treatments can be tested in clinical trials. However, their use in developing new therapies for sepsis has been controversial because of limitations of the models and inconsistencies with the clinical conditions. In consideration of the revised definition for clinical sepsis and septic shock (Sepsis-3), a Wiggers-Bernard Conference was held in Vienna in May 2017 to propose standardized guidelines on preclinical sepsis modeling. The participants conducted a literature review of 260 most highly cited scientific articles on sepsis models published between 2003 and 2012. The review showed, for example, that mice were used in 79% and euthanasia criteria were defined in 9% of the studies. Part I of this report details the recommendations for study design and humane modeling endpoints that should be addressed in sepsis models. The first recommendation is that survival follow-up should reflect the clinical time course of the infectious agent used in the sepsis model. Furthermore, it is recommended that therapeutic interventions should be initiated after the septic insult replicating clinical care. To define an unbiased and reproducible association between a new treatment and outcome, a randomization and blinding of treatments as well as inclusion of all methodological details in scientific publications is essential. In all preclinical sepsis studies, the high standards of animal welfare must be implemented. Therefore, development and validation of specific criteria for monitoring pain and distress, and euthanasia of septic animals, as well as the use of analgesics are recommended. A set of four considerations is also proposed to enhance translation potential of sepsis models. Relevant biological variables and comorbidities should be included in the study design and sepsis modeling should be extended to mammalian species other than rodents. In addition, the need for source control (in case of a defined infection focus) should be considered. These recommendations and considerations are proposed as "best practices" for animal models of sepsis that should be implemented.
Collapse
Affiliation(s)
- Basilia Zingarelli
- Department of Pediatrics, Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, College of Medicine, University of Cincinnati, Cincinnati, Ohio
| | | | - Susanne Drechsler
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology in the AUVA Research Center, Vienna, Austria
| | - Philip Efron
- Sepsis and Critical Illness Research Center, University of Florida College of Medicine, Gainesville, Florida
| | - John C Marshall
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, University of Toronto, Toronto, Canada
| | - Lyle Moldawer
- Sepsis and Critical Illness Research Center, University of Florida College of Medicine, Gainesville, Florida
| | - W Joost Wiersinga
- Division of Infectious Diseases, Center for Experimental and Molecular Medicine, The Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Xianzhong Xiao
- Xiangya School of Medicine, Central South University, Chagnsha, Hunan, China
| | - Marcin F Osuchowski
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology in the AUVA Research Center, Vienna, Austria
| | - Christoph Thiemermann
- The William Harvey Research Institute, Barts and London School of Medicine & Dentistry, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
8
|
Ge Y, Huang M, Yao YM. Recent advances in the biology of IL-1 family cytokines and their potential roles in development of sepsis. Cytokine Growth Factor Rev 2018; 45:24-34. [PMID: 30587411 DOI: 10.1016/j.cytogfr.2018.12.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 12/16/2018] [Accepted: 12/19/2018] [Indexed: 12/13/2022]
Abstract
The IL-1 family comprises two anti-inflammatory cytokines (IL-37, IL-38), two receptor antagonists (IL-1ra, IL-36ra), and seven ligand agonists (IL-1α, IL-1β, IL-33, IL-36α, IL-36β, IL-36γ). The members of this family exert pleiotropic effects on intercellular signaling, leading to pro- or anti-inflammatory responses. They initiate potent inflammatory and immune responses by binding to specific receptors in the IL-1 receptor family, and their activities are repressed by naturally occurring inhibitors. Various immune cells produce and are regulated by these crucial molecules, which appear to be involved in the pathogenesis of diverse diseases including cancer as well as inflammatory and autoimmune disorders. Recent decades have seen substantial progress in understanding how the IL-1 family contributes to the development of sepsis. In this review, we will briefly introduce the IL-1 family and discuss its critical role in inflammatory and immune responses. The potential significance of IL-1 members in sepsis will also be explored, together with the clinical implications for treating this dangerous condition.
Collapse
Affiliation(s)
- Yun Ge
- Department of General Intensive Care Unit, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310052, China
| | - Man Huang
- Department of General Intensive Care Unit, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310052, China
| | - Yong-Ming Yao
- Trauma Research Center, Fourth Medical Center of the Chinese PLA General Hospital, Beijing 100048, China.
| |
Collapse
|
9
|
Sepsis Pathophysiology, Chronic Critical Illness, and Persistent Inflammation-Immunosuppression and Catabolism Syndrome. Crit Care Med 2017; 45:253-262. [PMID: 27632674 DOI: 10.1097/ccm.0000000000002074] [Citation(s) in RCA: 331] [Impact Index Per Article: 41.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVES To provide an appraisal of the evolving paradigms in the pathophysiology of sepsis and propose the evolution of a new phenotype of critically ill patients, its potential underlying mechanism, and its implications for the future of sepsis management and research. DESIGN Literature search using PubMed, MEDLINE, EMBASE, and Google Scholar. MEASUREMENTS AND MAIN RESULTS Sepsis remains one of the most debilitating and expensive illnesses, and its prevalence is not declining. What is changing is our definition(s), its clinical course, and how we manage the septic patient. Once thought to be predominantly a syndrome of over exuberant inflammation, sepsis is now recognized as a syndrome of aberrant host protective immunity. Earlier recognition and compliance with treatment bundles has fortunately led to a decline in multiple organ failure and in-hospital mortality. Unfortunately, more and more sepsis patients, especially the aged, are suffering chronic critical illness, rarely fully recover, and often experience an indolent death. Patients with chronic critical illness often exhibit "a persistent inflammation-immunosuppression and catabolism syndrome," and it is proposed here that this state of persisting inflammation, immunosuppression and catabolism contributes to many of these adverse clinical outcomes. The underlying cause of inflammation-immunosuppression and catabolism syndrome is currently unknown, but there is increasing evidence that altered myelopoiesis, reduced effector T-cell function, and expansion of immature myeloid-derived suppressor cells are all contributory. CONCLUSIONS Although newer therapeutic interventions are targeting the inflammatory, the immunosuppressive, and the protein catabolic responses individually, successful treatment of the septic patient with chronic critical illness and persistent inflammation-immunosuppression and catabolism syndrome may require a more complementary approach.
Collapse
|
10
|
Cho W, Koo JY, Park Y, Oh K, Lee S, Song JS, Bae MA, Lim D, Lee DS, Park SB. Treatment of Sepsis Pathogenesis with High Mobility Group Box Protein 1-Regulating Anti-inflammatory Agents. J Med Chem 2016; 60:170-179. [DOI: 10.1021/acs.jmedchem.6b00954] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Wansang Cho
- CRI
Center for Chemical Proteomics, Department of Chemistry, Seoul National University, Seoul 08826, Korea
| | - Ja Young Koo
- CRI
Center for Chemical Proteomics, Department of Chemistry, Seoul National University, Seoul 08826, Korea
| | - Yeonju Park
- Department
of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Keunhee Oh
- Transplantation
Research Institute, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Sanghee Lee
- CRI
Center for Chemical Proteomics, Department of Chemistry, Seoul National University, Seoul 08826, Korea
| | - Jin-Sook Song
- Korea
Bio-Organic Science Division, Korea Research Institute of Chemical Technology, Daejeon, 34114, Korea
| | - Myung Ae Bae
- Korea
Bio-Organic Science Division, Korea Research Institute of Chemical Technology, Daejeon, 34114, Korea
| | - Donghyun Lim
- Department
of Biophysics and Chemical Biology, Seoul National University, Seoul 08826, Korea
| | - Dong-Sup Lee
- Department
of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Seung Bum Park
- CRI
Center for Chemical Proteomics, Department of Chemistry, Seoul National University, Seoul 08826, Korea
- Department
of Biophysics and Chemical Biology, Seoul National University, Seoul 08826, Korea
| |
Collapse
|
11
|
Kutyrev I, Cleveland B, Leeds T, Wiens GD. Proinflammatory cytokine and cytokine receptor gene expression kinetics following challenge with Flavobacterium psychrophilum in resistant and susceptible lines of rainbow trout (Oncorhynchus mykiss). FISH & SHELLFISH IMMUNOLOGY 2016; 58:542-553. [PMID: 27693200 DOI: 10.1016/j.fsi.2016.09.053] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 09/22/2016] [Accepted: 09/26/2016] [Indexed: 06/06/2023]
Abstract
Flavobacterium psychrophilum (Fp) is the causative agent of bacterial cold water disease (BCWD) which causes appreciable economic losses in rainbow trout aquaculture. We previously reported development of a genetic line, designated ARS-Fp-R that exhibits higher survival relative to a susceptible line, designated ARS-Fp-S, following either laboratory or natural on-farm challenge. The objectives of this study were to determine the temporal kinetics of gene expression between experimentally-challenged ARS-Fp-R and ARS-Fp-S fish and the correlation between gene expression and pathogen load. We developed a GeXP multiplex RT-PCR assay to simultaneously examine expression of immune-relevant genes, concentrating on tumor necrosis factor and interleukin-1 ligand/receptor systems and acute phase response genes. Spleen tissue was sampled at 6 h, 24 h, 48 h and 144 h post-challenge and pathogen load quantified by qPCR. Transcript abundance of cytokine genes tnfa1, tnfa2, tnfa3, il1b1, il1b2, il11a; acute phase response genes saa and drtp1; and putative cytokine receptors il1r1-like-b, il1r2, tnfrsf1a, tnfrsf9, tnfrsf1a-like-b increased following challenge while the transcript abundance of il1r-like-1 and tnfrsf1a-like-a decreased compared to PBS-injected line-matched control fish. Principal component analysis identified transcript levels of genes il1r-like-1 and tnfrsf1a-like-a as exhibiting differential expression between genetic lines. In summary, Fp i.p. injection challenge elicited a proinflammatory cytokine gene expression response in the spleen, with ARS-Fp-R line fish exhibiting modestly higher basal expression levels of several putative cytokine receptors. This study furthers the understanding of the immune response following Fp challenge and differences in gene expression associated with selective breeding for disease resistance.
Collapse
Affiliation(s)
- Ivan Kutyrev
- National Center for Cool and Cold Water Aquaculture, Agricultural Research Service, USDA, 11861 Leetown Rd, Kearneysville, WV 25430, USA; Institute of General and Experimental Biology, Siberian Branch of Russian Academy of Sciences, Sakhyanovoi St., 6, 670047 Ulan-Ude, Russia.
| | - Beth Cleveland
- National Center for Cool and Cold Water Aquaculture, Agricultural Research Service, USDA, 11861 Leetown Rd, Kearneysville, WV 25430, USA
| | - Timothy Leeds
- National Center for Cool and Cold Water Aquaculture, Agricultural Research Service, USDA, 11861 Leetown Rd, Kearneysville, WV 25430, USA
| | - Gregory D Wiens
- National Center for Cool and Cold Water Aquaculture, Agricultural Research Service, USDA, 11861 Leetown Rd, Kearneysville, WV 25430, USA.
| |
Collapse
|
12
|
Camerota A, Lögdberg L, Lake P, Larkin V, Fink M. Delayed therapy with a polymyxin B-dextran conjugate (PMX-622) improves survival in rabbits with Gram-negative peritonitis. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/096805199700400406] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Polymyxin B (PMB) is an amphipathic nephrotoxic antibiotic, which has been shown to neutralize the effects of endotoxin both in vitro and in vivo. PMB-D70 (PMX-622), a covalent conjugate of PMB with dextran 70 (D70), is less nephrotoxic than the parent compound. We sought to determine whether therapy with PMB-D70, in addition to conventional antimicrobial chemotherapy, could improve survival in a model of Gram-negative peritonitis. At T = 0 h, New Zealand white rabbits were implanted intraperitoneally with 10 ml of a suspension containing hemoglobin (40 μg/ml), mucin (150 μg/ml), and 1.0 ± 0.2 x 104 cfu/kg of viable Escherichia coli (O18:K1). Beginning at T = 4 h, the rabbits were treated with gentamicin (5 mg/kg every 12 h) for five doses or until death, and infused for 24 h or until death with either D70 or PMB-D70. Two pairs of groups were studied (doses indicate cumulative amounts infused over 24 h). The PMB-D70 (low dose) group received PMB-D70 (5 mg/kg of the PMB component) and the D70 (low dose) group received an equivalent dose of D70. The PMB-D70 (high dose) group received PMB-D70 (10 mg/kg of the PMB component) and the D70 (high dose) group received an equivalent dose of D70. Results for the two PMB-D70 groups, on the one hand, and the two D70 group, on the other hand, were statistically indistiguishable and, accordingly, were pooled for all analyses. Survival at 7 days was 11/25 (44%) for rabbits treated with PMB-D70 as compared to 2/23 (9%) for animals treated with D70 ( P = 0.007). We conclude that adjuvant treatment with PMB-D70 improves survival in a clinically relevant model of Gram-negative sepsis.
Collapse
Affiliation(s)
- A.J. Camerota
- Departments of Surgery and Anesthesiology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston Massachusetts, USA
| | - L. Lögdberg
- Sandoz Research Institute, East Hanover, New Jersey, USA
| | - P. Lake
- Sandoz Research Institute, East Hanover, New Jersey, USA
| | - V.A. Larkin
- Departments of Surgery and Anesthesiology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston Massachusetts, USA
| | - M.P. Fink
- Departments of Surgery and Anesthesiology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston Massachusetts, USA
| |
Collapse
|
13
|
Brand JD, Mathews JA, Kasahara DI, Wurmbrand AP, Shore SA. Regulation of IL-17A expression in mice following subacute ozone exposure. J Immunotoxicol 2016; 13:428-38. [PMID: 27043160 DOI: 10.3109/1547691x.2015.1120829] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Exposure to subacute ozone (O3) causes pulmonary neutrophil recruitment. In mice, this recruitment requires IL-17A. Ozone also causes expression of IL-23 and IL-1, which can induce IL-17A. The purpose of this study was to examine the hypothesis that IL-23 and IL-1 contribute to IL-17A expression and subsequent neutrophil recruitment after subacute O3 exposure. Wild-type, IL-23(-/-), and Flt3l(-/-) mice were exposed to air or 0.3 ppm O3 for 72 h. Flt3l(-/-) mice lack conventional dendritic cells (cDC) that can express IL-23 and IL-1. Other wild-type mice were pre-treated with saline or the IL-1R1 antagonist anakinra prior to O3 exposure. After exposure, bronchoalveolar lavage (BAL) was performed and lung tissue harvested. The results indicated that pulmonary Il17a mRNA abundance and IL-17A(+) F4/80(+) cells were significantly reduced in O3-exposed IL-23(-/-) vs in wild-type mice. In contrast, anakinra had no effect on Il23a or Il17a pulmonary mRNA abundance or on BAL concentrations of the neutrophil survival factor G-CSF, but anakinra did reduce BAL neutrophil numbers, likely because anakinra also reduced BAL IL-6. Compared to air, O3 caused a significant increase in DC numbers in wild-type, but not in Flt3(-/-) mice. However, there was no significant difference in Il23a or Il17a mRNA abundance or in BAL neutrophil count in O3-exposed Flt3(-/-) vs in wild-type mice. From these results, it was concluded that IL-23 but not IL-1 contributes to the IL-17A expression induced by subacute O3 exposure. Induction of IL-23 by O3 does not appear to require cDC.
Collapse
Affiliation(s)
- Jeffrey D Brand
- a Molecular and Integrative Physiological Sciences Program, Department of Environmental Health , Harvard T.H. Chan School of Public Health , Boston , MA , USA
| | - Joel A Mathews
- a Molecular and Integrative Physiological Sciences Program, Department of Environmental Health , Harvard T.H. Chan School of Public Health , Boston , MA , USA
| | - David I Kasahara
- a Molecular and Integrative Physiological Sciences Program, Department of Environmental Health , Harvard T.H. Chan School of Public Health , Boston , MA , USA
| | - Alison P Wurmbrand
- a Molecular and Integrative Physiological Sciences Program, Department of Environmental Health , Harvard T.H. Chan School of Public Health , Boston , MA , USA
| | - Stephanie A Shore
- a Molecular and Integrative Physiological Sciences Program, Department of Environmental Health , Harvard T.H. Chan School of Public Health , Boston , MA , USA
| |
Collapse
|
14
|
Kang JW, Kim SJ, Cho HI, Lee SM. DAMPs activating innate immune responses in sepsis. Ageing Res Rev 2015; 24:54-65. [PMID: 25816752 DOI: 10.1016/j.arr.2015.03.003] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Revised: 03/09/2015] [Accepted: 03/13/2015] [Indexed: 12/11/2022]
Abstract
Sepsis refers to the deleterious and non-resolving systemic inflammatory response of the host to microbial infection and is the leading cause of death in intensive care units. The pathogenesis of sepsis is highly complex. It is principally attributable to dysregulation of the innate immune system. Damage-associated molecular patterns (DAMPs) are actively secreted by innate immune cells and/or released passively by injured or damaged cells in response to infection or injury. In the present review, we highlight emerging evidence that supports the notion that extracellular DAMPs act as crucial proinflammatory danger signals. Furthermore, we discuss the potential of a wide array of DAMPs as therapeutic targets in sepsis.
Collapse
Affiliation(s)
- Jung-Woo Kang
- School of Pharmacy, Sungkyunkwan University, Seobu-ro 2066, Jangan-gu, Suwon, Gyeonggi-do, 440-746 South Korea
| | - So-Jin Kim
- School of Pharmacy, Sungkyunkwan University, Seobu-ro 2066, Jangan-gu, Suwon, Gyeonggi-do, 440-746 South Korea
| | - Hong-Ik Cho
- School of Pharmacy, Sungkyunkwan University, Seobu-ro 2066, Jangan-gu, Suwon, Gyeonggi-do, 440-746 South Korea
| | - Sun-Mee Lee
- School of Pharmacy, Sungkyunkwan University, Seobu-ro 2066, Jangan-gu, Suwon, Gyeonggi-do, 440-746 South Korea.
| |
Collapse
|
15
|
Franco R, Fernández-Suárez D. Alternatively activated microglia and macrophages in the central nervous system. Prog Neurobiol 2015; 131:65-86. [PMID: 26067058 DOI: 10.1016/j.pneurobio.2015.05.003] [Citation(s) in RCA: 495] [Impact Index Per Article: 49.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 05/22/2015] [Accepted: 05/30/2015] [Indexed: 12/20/2022]
Abstract
Macrophages are important players in the fight against viral, bacterial, fungal and parasitic infections. From a resting state they may undertake two activation pathways, the classical known as M1, or the alternative known as M2. M1 markers are mostly mediators of pro-inflammatory responses whereas M2 markers emerge for resolution and cleanup. Microglia exerts in the central nervous system (CNS) a function similar to that of macrophages in the periphery. Microglia activation and proliferation occurs in almost any single pathology affecting the CNS. Often microglia activation has been considered detrimental and drugs able to stop microglia activation were considered for the treatment of a variety of diseases. Cumulative evidence shows that microglia may undergo the alternative activation pathway, express M2-type markers and contribute to neuroprotection. This review focuses on details about the role of M2 microglia and in the approaches available for its identification. Approaches to drive the M2 phenotype and data on its potential in CNS diseases are also reviewed.
Collapse
Affiliation(s)
- Rafael Franco
- Molecular Neurobiology Laboratory, Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Barcelona, Barcelona, Spain; Centro Investigación Biomédica en Red: Enfermedades Neurodegenerativas (CIBERNED), Spain.
| | - Diana Fernández-Suárez
- Division of Molecular Neurobiology, Department of Neuroscience, Karolinska Institute, 17177 Stockholm, Sweden.
| |
Collapse
|
16
|
Ullewar MP, Umathe SN. A possible role of endogenous central corticotrophin releasing factor in lipopolysaccharide induced thymic involution and cell apoptosis: effect of peripheral injection of corticotrophin releasing factor. J Neuroimmunol 2015; 280:58-65. [PMID: 25773157 DOI: 10.1016/j.jneuroim.2015.03.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2014] [Revised: 12/12/2014] [Accepted: 03/01/2015] [Indexed: 10/23/2022]
Abstract
The aim of the study was to investigate the role of endogenous peripheral and central corticotrophin-releasing factor (CRF) following lipopolysaccharide (LPS) challenge on thymic involution and apoptosis. Administration of LPS (100 μg/mouse, ip) led to thymic involution, to a decrease of CD4+CD8+ thymocyte subset, and to fragmentation of thymic DNA. Pretreatment of LPS challenged mice with intracerebroventricular α-helical CRF (a CRF antagonist) attenuated the effect of LPS however, intraventricular administered α-helical CRF failed to affect LPS response on thymus. Moreover, the effects of LPS on thymus, examined on 1, 7 and 14 days were wholly abrogated by prior administration of intraventricular CRF (10 μg/animal). The plasma corticosterone levels were found to be decreased with single dose of peripheral CRF in LPS challenged mice. These findings indicate that central endogenous CRF involved in LPS induced thymic atrophy. However, peripheral CRF offers protective effect on LPS induced thymic involution and cell apoptosis.
Collapse
Affiliation(s)
- Meenal P Ullewar
- University Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj, Nagpur University, Mahatma Jyotiba Fuley Shaikshanik Parisar, Amravati Road, Nagpur 440 033 MS, India
| | - Sudhir N Umathe
- Kamla Nehru College of Pharmacy, Borkhedi Gate, Near Railway Crossing, Butibori, Nagpur, 441108 MS, India.
| |
Collapse
|
17
|
Abstract
Sepsis, a common and potentially fatal systemic illness, is triggered by microbial infection and often leads to impaired function of the lungs, kidneys or other vital organs. Since the early 1980s, a large number of therapeutic agents for the treatment of sepsis have been evaluated in randomized controlled clinical trials. With few exceptions, the results from these trials have been disappointing, and no specific therapeutic agent is currently approved for the treatment of sepsis. To improve upon this dismal record, investigators will need to identify more suitable therapeutic targets, improve their approaches for selecting candidate compounds for clinical development and adopt better designs for clinical trials.
Collapse
Affiliation(s)
- Mitchell P Fink
- Departments of Surgery and Anesthesiology, David Geffen School of Medicine at University of California, Los Angeles, 10833 Le Conte Avenue, 72-160 CHS, Los Angeles California 90095, USA
| | - H Shaw Warren
- Infectious Disease Units, Departments of Pediatrics and Medicine, Massachusetts General Hospital East, 149 13th Street, Fifth Floor, Charlestown, Massachusetts 02129, USA
| |
Collapse
|
18
|
Censoplano N, Epting CL, Coates BM. The Role of the Innate Immune System in Sepsis. CLINICAL PEDIATRIC EMERGENCY MEDICINE 2014. [DOI: 10.1016/j.cpem.2014.04.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
19
|
Abstract
Sepsis remains a common, serious, and heterogeneous clinical entity that is difficult to define adequately. Despite its importance as a public health problem, efforts to develop and gain regulatory approval for a specific therapeutic agent for the adjuvant treatment of sepsis have been remarkably unsuccessful. One step in the critical pathway for the development of a new agent for adjuvant treatment of sepsis is evaluation in an appropriate animal model of the human condition. Unfortunately, the animal models that have been used for this purpose have often yielded misleading findings. It is likely that there are multiple reasons for the discrepancies between the results obtained in tests of pharmacological agents in animal models of sepsis and the outcomes of human clinical trials. One of important reason may be that the changes in gene expression, which are triggered by trauma or infection, are different in mice, a commonly used species for preclinical testing, and humans. Additionally, many species, including mice and baboons, are remarkably resistant to the toxic effects of bacterial lipopolysaccharide, whereas humans are exquisitely sensitive. New approaches toward the use of animals for sepsis research are being investigated. But, at present, results from preclinical studies of new therapeutic agents for sepsis must be viewed with a degree of skepticism.
Collapse
Affiliation(s)
- Mitchell P Fink
- Departments of Surgery and Anesthesiology; David Geffen School of Medicine at UCLA; Los Angeles, CA USA
| |
Collapse
|
20
|
|
21
|
Ikeda M, Ohira H, Toyama Y, Katagiri T, Sakakibara B. Effect of intestinal microflora on the production of interleukin 10 and prostaglandin e(2) in serum and kupffer cells from germfree and conventional mice. J Clin Biochem Nutr 2011; 41:169-74. [PMID: 18299711 PMCID: PMC2243250 DOI: 10.3164/jcbn.2007023] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2006] [Accepted: 04/02/2007] [Indexed: 11/25/2022] Open
Abstract
To determine why germfree (GF) mice are less productivity of proinflammatory cytokines than conventional (CV) mice, we studied serum levels of interleukin 10 (IL-10) and prostaglandin E2 (PGE2) in mice after treatment with lipopolyssacharide (LPS). A single injection of LPS caused an elevation of IL-10 in serum from GF, LPS-GF (germfree mice given drinking water containing LPS) and CV mice. The response was highest in serum from GF mice, and was lower in serum from LPS-GF mice compared with GF mice. Before LPS injection, serum PGE2 was significantly higher in CV and LPS-GF mice than in GF ones. After LPS injection, a higher level of PGE2 was maintained over 12 h in CV mice after LPS injection, while the LPS treatment reduced the level in LPS-GF mice and increased the level in GF mice. The levels of IL-10 in culture medium from Kupffer cells treated with LPS showed similar results to serum in GF and CV mice. These results suggest that high levels of IL-10 in serum from germfree mice may be partly responsible for the lower in vivo responsiveness of these proinflammatory cytokines to LPS in these mice, although PGE2 was not responsible for the lower responsiveness of these inflammatory cytokines to LPS.
Collapse
Affiliation(s)
- Masamichi Ikeda
- Laboratory of Nutritional Pathology, Faculty of Nutrition, Kobegakuin University Ikawadani-cho, Nishi-ku, Kobe 651-2180, Japan
| | | | | | | | | |
Collapse
|
22
|
Berry CA, Nitsos I, Hillman NH, Pillow JJ, Polglase GR, Kramer BW, Kemp MW, Newnham JP, Jobe AH, Kallapur SG. Interleukin-1 in lipopolysaccharide induced chorioamnionitis in the fetal sheep. Reprod Sci 2011; 18:1092-102. [PMID: 21493953 DOI: 10.1177/1933719111404609] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
We tested the hypothesis that interleukin 1 (IL-1) mediates intra-amniotic lipopolysaccharide (LPS)-induced chorioamnionitis in preterm fetal sheep. Time-mated Merino ewes with singleton fetuses received IL-1α, LPS, or saline (control) by intra-amniotic injection 1 to 2 days before operative delivery at 124 ± 1 days gestational age (N = 5-9/group; term = 150 days). Recombinant human IL-1 receptor antagonist (rhIL-1ra) was given into the amniotic fluid 3 hours before intra-amniotic LPS or saline to block IL-1 signaling. Inflammation in the chorioamnion was determined by histology, cytokine messenger RNA (mRNA), protein expression, and by quantitation of activated inflammatory cells. Intra-amniotic IL-1 and LPS both induced chorioamnionitis. However, IL-1 blockade with IL-1ra did not decrease intra-amniotic LPS-induced increases in pro-inflammatory cytokine mRNAs, numbers of inflammatory cells, myeloperoxidase, or monocyte chemotactic protein-1-expressing cells in the chorioamnion. We conclude that IL-1 and LPS both can cause chorioamnionitis, but IL-1 is not an important mediator of LPS-induced chorioamnionitis in fetal sheep.
Collapse
Affiliation(s)
- Clare A Berry
- Division of Pulmonary Biology/Neonatology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Herold S, Tabar TS, Janssen H, Hoegner K, Cabanski M, Lewe-Schlosser P, Albrecht J, Driever F, Vadasz I, Seeger W, Steinmueller M, Lohmeyer J. Exudate macrophages attenuate lung injury by the release of IL-1 receptor antagonist in gram-negative pneumonia. Am J Respir Crit Care Med 2011; 183:1380-90. [PMID: 21278303 DOI: 10.1164/rccm.201009-1431oc] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Exudate macrophages are key players in host defense toward invading pathogens. Their antiinflammatory and epithelial-protective potential in gram-negative pneumonia, however, remains elusive. OBJECTIVES We investigated whether exudate macrophages contributed to preservation of alveolar epithelial barrier integrity and analyzed the molecular pathways involved. METHODS We evaluated the antiinflammatory and epithelial-protective effects of exudate macrophages in a model of LPS- and Klebsiella pneumoniae-induced lung injury comparing wild-type and CC-chemokine receptor 2 (CCR2)-deficient mice with defective lung macrophage recruitment and in in vitro studies using primary alveolar epithelial cells. MEASUREMENTS AND MAIN RESULTS CCR2(-/-) mice exhibited enhanced alveolar epithelial cell apoptosis and lung leakage on intratracheal LPS treatment, which could be attributed to lack of exudate macrophage recruitment from the circulating pool as demonstrated in a model of wild-type/CCR2(-/-) bone-marrow chimeric mice. Among various antiinflammatory and proliferative mediators analyzed, the endogenous counterpart of resident macrophage-expressed IL-1β, IL-1 receptor antagonist (IL-1ra), was highly up-regulated in flow-sorted exudate macrophages in LPS-treated wild-type mice. LPS/IL-1β-induced impairment of alveolar epithelial cell integrity was antagonized by IL-1ra in vitro. Finally, intratracheal substitution of IL-1ra or intravenous adoptive transfer of IL-1ra(+/+) but not IL-1ra(-/-) blood mononuclear cells attenuated alveolar inflammation, epithelial apoptosis, and loss of barrier function in LPS-challenged or K. pneumoniae-infected CCR2(-/-) mice and enhanced survival after K. pneumoniae infection. CONCLUSIONS We conclude that recruited lung macrophages attenuate IL-1β-mediated acute lung injury in gram-negative pneumonia by release of IL-1ra.
Collapse
Affiliation(s)
- Susanne Herold
- Department of Internal Medicine II, University of Giessen Lung Center, Giessen, Germany.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Lamacchia C, Palmer G, Bischoff L, Rodriguez E, Talabot-Ayer D, Gabay C. Distinct Roles of Hepatocyte- and Myeloid Cell-Derived IL-1 Receptor Antagonist during Endotoxemia and Sterile Inflammation in Mice. THE JOURNAL OF IMMUNOLOGY 2010; 185:2516-24. [DOI: 10.4049/jimmunol.1000872] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
25
|
The 2008 ESPEN Sir David Cuthbertson Lecture: Fatty acids and inflammation--from the membrane to the nucleus and from the laboratory bench to the clinic. Clin Nutr 2010; 29:5-12. [PMID: 19931231 DOI: 10.1016/j.clnu.2009.11.003] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2009] [Revised: 09/02/2009] [Accepted: 11/04/2009] [Indexed: 11/21/2022]
Abstract
Many chronic conditions involve excessive inflammation that is damaging to host tissues. Excessive or inappropriate inflammation and immunosuppression are components of the response to surgery, trauma, injury and infection in some individuals and these can lead, progressively, to sepsis and septic shock. Hyperinflammation is characterised by the production of inflammatory cytokines, eicosanoids and other inflammatory mediators, while the immunosuppression is characterised by impairment of antigen presentation and of certain T cell responses. N-6 fatty acids may contribute to the hyperinflamed and immunosuppressed states. N-3 fatty acids from fish oil decrease the production of inflammatory cytokines and eicosanoids. They act both directly (by replacing arachidonic acid as an eicosanoid precursor) and indirectly (by altering the expression of inflammatory genes through effects on transcription factor activation). Thus, these fatty acids are potentially useful anti-inflammatory agents and may be of benefit in patients with chronic inflammatory diseases or at risk of hyperinflammation and sepsis. An emerging application of n-3 fatty acids is in surgical or critically ill patients where they may be added to parenteral or enteral formulas. Studies to date are suggestive of clinical benefits from these approaches, although more robust data are needed especially in critically ill patients.
Collapse
|
26
|
Zaki MES, Elgendy MY, El-Mashad NBED, Farahat ME. IL-18 Level Correlates with Development of Sepsis in Surgical Patients. Immunol Invest 2009; 36:403-11. [PMID: 17691022 DOI: 10.1080/08820130701244275] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Interleukin 18 may play an important role in sepsis. We measured circulating IL-18 levels in 47 patients with surgical wounds, 27 of them had clinical and laboratory evidences of sepsis. The patients had significantly higher IL-18 levels than healthy control (p1 = 0.001), patients with local infection (p4 = 0.001) and patients with clean surgical wound (p5 = 0.001) both at admission and after 48 hours. Serial observations revealed that in all patients, IL-18 levels increased significantly especially in patients with sepsis after 48 h (p = 0.007). IL-18 levels appeared to correlate significantly with leucocytes counts (p = 0.001) and with C-RP (p = 0.007). These results suggest that IL-18 levels are increased in patients with sepsis and correlate with CRP and leukocyte counts suggesting that IL-18 may play a role in sepsis. We suggest that IL-18 could be used as a collaborative evidence of surgical sepsis.
Collapse
Affiliation(s)
- Maysaa El-Sayed Zaki
- Department of Clinical Pathology, Faculty of Medicine, Mansoura University, Mansoura, Egypt.
| | | | | | | |
Collapse
|
27
|
Kallapur SG, Nitsos I, Moss TJM, Polglase GR, Pillow JJ, Cheah FC, Kramer BW, Newnham JP, Ikegami M, Jobe AH. IL-1 mediates pulmonary and systemic inflammatory responses to chorioamnionitis induced by lipopolysaccharide. Am J Respir Crit Care Med 2009; 179:955-61. [PMID: 19234101 DOI: 10.1164/rccm.200811-1728oc] [Citation(s) in RCA: 106] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
RATIONALE Chorioamnionitis frequently associates with preterm delivery and increased amniotic fluid IL-1, and causes fetal lung and systemic inflammation. However, chorioamnionitis is also associated with a paradoxical reduction in the incidence of surfactant deficiency-related respiratory distress syndrome in preterm infants. OBJECTIVES To identify the role of IL-1 signaling in the mediation of pulmonary and systemic inflammation and lung maturation in a fetal sheep model of lipopolysaccharide (LPS) induced chorioamnionitis. METHODS After confirming the efficacy of recombinant human IL-1 receptor antagonist (rhIL-1ra), fetal sheep were exposed to intraamniotic (IA) injections of Escherichia coli LPS with or without prior IA injections of rhIL-1ra. Preterm lambs were delivered at 82% of term gestation. MEASUREMENTS AND MAIN RESULTS rhIL-1ra decreased IA LPS-induced lung inflammation assessed by decreased lung neutrophil and monocyte influx, inducible nitric oxide synthase expression, lung IL-6 and IL-1beta mRNA expression, and airway myeloperoxidase concentrations. rhIL-1ra inhibited IA LPS-induced fetal systemic inflammation assessed by decreased plasma IL-8, protein carbonyls, blood neutrophilia, and the expression of serum amyloid A3 mRNA in the liver. rhIL-1ra also partially blocked the lung maturational effects of IA LPS. Therefore blockade of IL-1 signaling in the amniotic compartment inhibited fetal lung and systemic inflammation and lung maturation in response to LPS-induced chorioamnionitis. CONCLUSIONS IL-1 plays a central role in the pathogenesis of chorioamnionitis-induced fetal inflammatory responses.
Collapse
Affiliation(s)
- Suhas G Kallapur
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Trakala M, Arias CF, García MI, Moreno-Ortiz MC, Tsilingiri K, Fernández PJ, Mellado M, Díaz-Meco MT, Moscat J, Serrano M, Martínez-A C, Balomenos D. Regulation of macrophage activation and septic shock susceptibilityviap21(WAF1/CIP1). Eur J Immunol 2009; 39:810-9. [DOI: 10.1002/eji.200838676] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
29
|
Gu CH, Cui Q, Wang YY, Wang J, Dou YW, Zhao R, Liu Y, Wang J, Pei JM, Yi DH. Effects of insulin therapy on inflammatory mediators in infants undergoing cardiac surgery with cardiopulmonary bypass. Cytokine 2008; 44:96-100. [DOI: 10.1016/j.cyto.2008.06.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2008] [Revised: 06/04/2008] [Accepted: 06/27/2008] [Indexed: 01/04/2023]
|
30
|
Lipopolysaccharide-Induced Upregulation of Tumor Necrosis Factor-α (TNF-α) in Rat Spinal Cord. Inflammation 2008; 31:336-43. [DOI: 10.1007/s10753-008-9083-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
31
|
Singer JW, Rursten SL, Rice GC, Perry Gordon W, Bianco JA. Inhibitors of intracellular phosphatidic acid production: novel therapeutics with broad clinical applications. Expert Opin Investig Drugs 2008. [DOI: 10.1517/13543784.3.6.631] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
32
|
Vincent JL. Editorial: Pulmonary-Allergy, Dermatological, Gastrointestinal & Arthritis: Clinical trials in immunotherapy for sepsis: Why have the results been so disappointing? Expert Opin Investig Drugs 2008. [DOI: 10.1517/13543784.5.1.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
33
|
Jeschke MG, Herndon DN. The Hepatic Response to Severe Injury. Intensive Care Med 2007. [DOI: 10.1007/978-0-387-49518-7_58] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
34
|
Leffler M, Hrach T, Stuerzl M, Horch RE, Herndon DN, Jeschke MG. Insulin attenuates apoptosis and exerts anti-inflammatory effects in endotoxemic human macrophages. J Surg Res 2007; 143:398-406. [PMID: 17583747 DOI: 10.1016/j.jss.2007.01.030] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2006] [Revised: 01/16/2007] [Accepted: 01/25/2007] [Indexed: 12/13/2022]
Abstract
BACKGROUND Insulin decreases the incidence of sepsis and improves mortality of critically ill patients. In endotoxemic as well as in thermally injured rats, insulin attenuates the systemic inflammatory response by decreasing the proinflammatory and increasing the antiinflammatory cascade. The aim of the present study was to determine the effects of insulin on cell survival, cell activity, apoptosis, and proinflammatory response in a human macrophage-like cell line (THP-1 cells) stressed with lipopolysaccharide (LPS). MATERIALS AND METHODS Human macrophages were stressed with LPS and received either saline or insulin. Cell viability was analyzed by MTS, apoptosis was detected using JC-1 and terminal deoxynucleotidyl transferase-mediated nick end labeling-staining, and to elucidate on the signaling pathway, we used wortmannin as a phosphatidylinositol-3-kinase inhibitor. Tumor necrosis factor (TNF) and interleukin-1beta (IL-1beta) were measured to determine the effect of insulin on proinflammatory cytokine expression. RESULTS Insulin caused a significant increase in cell viability and significantly reduced apoptosis in LPS-stimulated human macrophages in a dose-dependent manner. The antiapoptotic effect of insulin could be completely blocked with the addition of wortmannin. Insulin significantly decreased TNF and IL-1beta in endotoxemic human macrophages. CONCLUSIONS Our results indicate that insulin exerts antiapoptotic effects and reduces the expression of proinflammatory cytokines in endotoxemic human macrophages. The antiapoptotic effects are mediated via the phospatidylinositol-3-kinase-pathway.
Collapse
Affiliation(s)
- Mareike Leffler
- Department of Plastic and Hand Surgery, University Hospital Erlangen-Nürnberg, Erlangen, Germany
| | | | | | | | | | | |
Collapse
|
35
|
Stojanovic I, Cuzzocrea S, Mangano K, Mazzon E, Miljkovic D, Wang M, Donia M, Al Abed Y, Kim J, Nicoletti F, Stosic-Grujicic S, Claesson M. In vitro, ex vivo and in vivo immunopharmacological activities of the isoxazoline compound VGX-1027: Modulation of cytokine synthesis and prevention of both organ-specific and systemic autoimmune diseases in murine models. Clin Immunol 2007; 123:311-23. [PMID: 17449326 DOI: 10.1016/j.clim.2007.03.004] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2006] [Revised: 02/26/2007] [Accepted: 03/07/2007] [Indexed: 01/08/2023]
Abstract
We have presently studied the in vitro, ex vivo and in vivo immunopharmacological effects of VGX-1027 [(S,R)-3-phenyl-4,5-dihydro-5-isoxasole acetic acid]. This compound reduced the secretion of IL-1beta, TNF-alpha and IL-10 from purified murine macrophages stimulated "in vitro" with lipopolysaccharide (LPS), and it also modified the signaling pathways induced in these cells by LPS entailing reduced activation of NF-kappaB and p38 MAP kinase pathways along with up-regulation of ERK pathways. VGX-1027 appeared to spare T cell function as it was unable to modify the proliferation and/or secretion of IL-2, IFN-gamma and IL-4 induced in purified murine CD4+ T cells from stimulation with either CD3+CD28 or ConA. These effects on macrophages may account for the capacity of VGX-1027 to markedly ameliorate the course of both acute and chronic immunoinflammatory diseases in mice such as carrageenan-induced pleurisy, LPS-induced lethality and type II collagen-induced arthritis. Acute and subacute toxicological studies show that the drug is not toxic at the doses that exert biological effects in these preclinical models. These data warrant additional studies for the potential use of VGX-1027 in the clinical setting.
Collapse
MESH Headings
- Acetates/pharmacology
- Acetates/toxicity
- Acetylmuramyl-Alanyl-Isoglutamine/pharmacology
- Animals
- Arthritis, Experimental/chemically induced
- Arthritis, Experimental/pathology
- Arthritis, Experimental/prevention & control
- CD4-Positive T-Lymphocytes/drug effects
- CD4-Positive T-Lymphocytes/metabolism
- Cell Proliferation/drug effects
- Cell Survival/drug effects
- Cytokines/metabolism
- Female
- I-kappa B Proteins/metabolism
- Immunologic Factors/pharmacology
- Immunologic Factors/toxicity
- Leukocytes, Mononuclear/drug effects
- Leukocytes, Mononuclear/metabolism
- Lipopolysaccharides/pharmacology
- Macrophages, Peritoneal/drug effects
- Macrophages, Peritoneal/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Inbred CBA
- Mice, Inbred DBA
- Mitogen-Activated Protein Kinases/metabolism
- Oxazoles/pharmacology
- Oxazoles/toxicity
- Phosphorylation/drug effects
- Pleurisy/chemically induced
- Pleurisy/pathology
- Pleurisy/prevention & control
- Shock, Septic/chemically induced
- Shock, Septic/mortality
- Shock, Septic/prevention & control
- Spleen/cytology
- Spleen/drug effects
- Survival Rate
Collapse
Affiliation(s)
- Ivana Stojanovic
- Department of Immunology, Institute for Biological Research Sinisa Stankovic, Belgrade, Serbia and Montenegro
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
Excessive or inappropriate inflammation and immunosuppression are components of the response to surgery, trauma, injury and infection in some individuals and can lead, progressively, to sepsis and septic shock. The hyperinflammation is characterised by the production of inflammatory cytokines, arachidonic acid-derived eicosanoids and other inflammatory mediators, while the immunosuppression is characterised by impairment of antigen presentation and of T-helper lymphocyte type-1 responses. Long-chain n-3 fatty acids from fish oil decrease the production of inflammatory cytokines and eicosanoids. They act both directly (by replacing arachidonic acid as an eicosanoid substrate and by inhibiting arachidonic acid metabolism) and indirectly (by altering the expression of inflammatory genes through effects on transcription factor activation). Thus, long-chain n-3 fatty acids are potentially useful anti-inflammatory agents and may be of benefit in patients at risk of hyperinflammation and sepsis. As a consequence, an emerging application for n-3 fatty acids, in which they may be added to parenteral (or enteral) formulas, is in surgical or critically-ill patients. Parenteral nutrition that includes n-3 fatty acids appears to preserve immune function better than standard formulas and appears to diminish the extent of the inflammatory response. Studies to date are suggestive of clinical benefits from these approaches, especially in patients post surgery, although evidence of clinical benefit in patients with sepsis is emerging.
Collapse
Affiliation(s)
- Philip C Calder
- Institute of Human Nutrition, School of Medicine, University of Southampton, Bassett Crescent East, Southampton SO16 7PX, UK.
| |
Collapse
|
37
|
Inoue KI, Takano H, Sakurai M, Oda T, Tamura H, Yanagisawa R, Shimada A, Yoshikawa T. Pulmonary exposure to diesel exhaust particles enhances coagulatory disturbance with endothelial damage and systemic inflammation related to lung inflammation. Exp Biol Med (Maywood) 2006; 231:1626-32. [PMID: 17060683 DOI: 10.1177/153537020623101007] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Pulmonary exposure to diesel exhaust particles (DEP) enhances lung inflammation related to bacterial endotoxin (lipopolysaccharide [LPS]) in mice. Severe lung inflammation can reportedly induce coagulatory abnormalities and systemic inflammation. This study examined the effects of components of DEP on lung inflammation, pulmonary permeability, coagulatory changes, systemic inflammatory response, and lung-to-systemic translocation of LPS in a murine model of lung inflammation. ICR mice were divided into six experimental groups that intratracheally received vehicle, LPS (2.5 mg/kg), organic chemicals in DEP (DEP-OC; 4 mg/kg) extracted with dicloromethane), residual carbonaceous nuclei of DEP (washed DEP: 4 mg/kg), DEP-OC + LPS, or washed DEP + LPS. Both DEP components exacerbated lung inflammation, vascular permeability, and the increased fibrinogen and E-selectin levels induced by LPS. With overall trends, the exacerbation was more prominent with washed DEP than with DEP-OC. Washed DEP + LPS significantly decreased activated protein C and antithrombin-III and elevated circulatory levels of interleukin (IL)-6, keratinocyte chemoattractant (KC), and LPS as compared with LPS alone, whereas DEP-OC + LPS elevated IL-6, KC, and LPS without significance. These results show that DEP components, especially washed DEP, amplify the effects if LPS on the respiratory system and suggest that they contribute to the adverse health effects of particulate air pollution on the sensitive populations with predisposing vascular and/or pulmonary diseases, including ischemic vascular diseases and respiratory infection.
Collapse
Affiliation(s)
- Ken-Ichiro Inoue
- Environmental Health Sciences Division, National Institute for Environmental Studies, 16-2 Onogawa, Tsukuba 305-8506, Japan
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Kirisawa R, Hashimoto N, Tazaki M, Yamanaka H, Ishii R, Hagiwara K, Iwai H. Molecular characterization and functional expression of equine interleukin-1 type I and type II receptor cDNAs. Vet Immunol Immunopathol 2006; 109:219-31. [PMID: 16176839 DOI: 10.1016/j.vetimm.2005.08.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2005] [Revised: 07/05/2005] [Accepted: 08/12/2005] [Indexed: 11/15/2022]
Abstract
cDNA generated from lipopolysaccharide-stimulated equine peripheral blood mononuclear cells was used to amplify and clone type I and type II equine interleukin-1 receptors (IL-1RI and IL-1RII) using primers derived from semi-conserved regions between human and mouse IL-1RI and IL-1RII sequences, respectively. 5' and 3' terminal sequences of equine IL-1RI and IL-1RII were amplified by 5' and 3' rapid amplification of cDNA ends. The deduced amino acid sequence of equine IL-1RI demonstrated 77, 64 and 63% similarity with human, mouse and rat sequences, respectively. The predicted amino acid sequence of equine IL-1RII demonstrated 70, 60 and 58% similarity with human, mouse and rat sequences, respectively. Recombinant equine soluble IL-1RI and IL-1RII produced in insect cells bound recombinant equine IL-1alpha and IL-1beta. Furthermore, both receptors suppressed the growth inhibitory activities of equine IL-1alpha and IL-1beta toward A375 cells in a dose-dependent manner, indicating that the present equine IL-1RI and IL-1RII cDNA encodes biologically active proteins.
Collapse
Affiliation(s)
- Rikio Kirisawa
- Department of Veterinary Microbiology, School of Veterinary Medicine, Rakuno Gakuen University, Bunkyoudai-Midorimachi 582, Ebetsu, Hokkaido 069-8501, Japan.
| | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
This review reviews the critical role played by cytokines in the pathogenesis of Escherichia coli sepsis. It focuses on prototypic pro-inflammatory and anti-inflammatory cytokines and their influence on mortality in experimental animal models of E. coli endotoxemia and of live E. coli sepsis. The review reviews the results of clinical trials on anticytokine therapy in patients with severe sepsis or septic shock. The recognition of the critical role played by tumor necrosis factor (TNF), a secreted 17kDa cytokine, in endotoxic and gram-negative shock has been a major step forward in our understanding of the pathogenesis of sepsis. The review describes the role of TNF, IL1, and IL6 in animal models of E. coli endotoxemia and sepsis. Given the pivotal role played by TNF in experimental sepsis and the fact that elevated concentrations of TNF were detected in the circulation of patients with sepsis, anti-TNF treatment strategies were investigated as adjunctive therapy for severe sepsis and septic shock. Several studies demonstrated that high levels of interleukin-6 (IL-6) are associated with an increased risk for fatal outcome. Gamma interferon (IFN-γ), IL-12, and IL-18 are functionally related cytokines. A recent study has indicated that transgenic mice overexpressing IL-15 are resistant to an otherwise lethal intraperitoneal E. coli challenge. IL4, IL10, and IL13are prototypic anti-inflammatory cytokines. Their classification as anti-inflammatory cytokines is based on the observation that these molecules inhibit the production of proinflammatory cytokines (primarily TNF and IL1) and toxic oxygen and reactive nitrogen species by myeloid cells.
Collapse
|
40
|
Turnbull IR, McDunn JE, Takai T, Townsend RR, Cobb JP, Colonna M. DAP12 (KARAP) amplifies inflammation and increases mortality from endotoxemia and septic peritonitis. ACTA ACUST UNITED AC 2005; 202:363-9. [PMID: 16061725 PMCID: PMC2213081 DOI: 10.1084/jem.20050986] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
DAP12 (KARAP) is a transmembrane signaling adaptor for a family of innate immunoreceptors that have been shown to activate granulocytes and monocytes/macrophages, amplifying production of inflammatory cytokines. Contrasting with these data, recent studies suggest that DAP12 signaling has an inhibitory role in the macrophage response to microbial products (Hamerman, J.A., N.K. Tchao, C.A. Lowell, and L.L. Lanier. 2005. Nat. Immunol. 6:579–586). To determine the in vivo role for DAP12 signaling in inflammation, we measured the response of wild-type (WT) and DAP12−/− mice to septic shock. We show that DAP12−/− mice have improved survival from both endotoxemia and cecal ligation and puncture–induced septic shock. As compared with WT mice, DAP12−/− mice have decreased plasma cytokine levels and a decreased acute phase response during sepsis, but no defect in the recruitment of cells or bacterial control. In cells isolated after sepsis and stimulated ex vivo, DAP12 signaling augments lipopolysaccharide-mediated cytokine production. These data demonstrate that, during sepsis, DAP12 signaling augments the response to microbial products, amplifying inflammation and contributing to mortality.
Collapse
Affiliation(s)
- Isaiah R Turnbull
- Department of Pathology and Immunology, Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110
| | | | | | | | | | | |
Collapse
|
41
|
Trepicchio WL, Dorner AJ. The therapeutic utility of Interleukin-11 in the treatment of inflammatory disease. Expert Opin Investig Drugs 2005; 7:1501-4. [PMID: 15992047 DOI: 10.1517/13543784.7.9.1501] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Interleukin-11 (IL-11) is a pleiotropic cytokine that exhibits anti-inflammatory and mucosal protective effects in a variety of animal models of acute and chronic inflammation, such as mucositis, inflammatory bowel disease and autoimmune joint disease. This reduction in inflammation and epithelial damage is mediated in part through effects of recombinant human (rh) IL-11 on macrophage effector function and epithelial cell growth. In vitro studies indicate that rhIL-11 inhibits tumour necrosis factor (TNF)-alpha, IL-1beta, IL-12, IL-6, and nitric oxide production from activated macrophages. Analysis of the effects of rhIL-11 on transcription factors that activate pro-inflammatory cytokines demonstrate that the level of induced nuclear factor kappa B (NF-kappaB) binding activity in the nucleus of rhIL-11-treated peritoneal macrophages is significantly reduced. Studies of normal intestinal epithelial cells indicate that rhIL-11 reduces the rate of cellular proliferation. Analysis of cell-cycle progression demonstrates that growth inhibition of epithelial cells by rhIL-11 correlates with delayed entry into S phase and suppression of pRB phosphorylation. IL-11 also protects intestinal crypt stem cells from radiation- or chemotherapy-induced insults. Such immunomodulatory and epithelial activities may contribute to the protective effects of this cytokine and support the clinical utility of rhIL-11 in the treatment of mucositis, as well as a variety of chronic inflammatory diseases, such as Crohn's disease and rheumatoid arthritis.
Collapse
Affiliation(s)
- W L Trepicchio
- Department of Preclinical Molecular and Cellular Biology, Genetics Institute, Andover, MA 01810, USA
| | | |
Collapse
|
42
|
Miyazaki S, Ishikawa F, Fujikawa T, Nagata S, Yamaguchi K. Intraperitoneal injection of lipopolysaccharide induces dynamic migration of Gr-1high polymorphonuclear neutrophils in the murine abdominal cavity. CLINICAL AND DIAGNOSTIC LABORATORY IMMUNOLOGY 2005; 11:452-7. [PMID: 15138169 PMCID: PMC404587 DOI: 10.1128/cdli.11.3.452-457.2004] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Intraperitoneal injection of lipopolysaccharide (LPS; 100 microg) in mice resulted in the disappearance of almost all proteose peptone-induced polymorphonuclear neutrophils (PMNs) with high-level fluorescence for the cell surface marker Gr-1 (Gr-1(high)) at 15 min postinjection, followed by doubling of their proportion at 30 min postinjection. High staining levels of 3'-acetyl-2'-carboxyl-6',7'-(dihyropyran-2'-one)-5 or 6-carboxyfluorescein diacethoxylmethyl ester-labeled PMNs injected into the peritoneal cavity were detected in mesenteric lymph nodes 15 min postinjection of LPS. Therefore, the time of decrease of Gr-1(high) PMNs coincided with that of the increase in cell accumulation in mesenteric lymph nodes. Since milk fat globule-EGF factor 8 (MFG-E8), which is secreted by macrophages, bound many PMNs exhibiting Gr-1(high) and Gr-1(medium) at 30 min postinjection of LPS, the staining level of annexin V on those cells was very low because its binding site is the same as the receptor for MFG-E8. At 60 min postinjection of LPS, the proportion of Gr-1(high) PMNs decreased, and almost all Gr-1(medium) PMNs tended to shift to the right compared with those at 30 min postinjection. The geomeans of Toll-like receptor 4 (TLR4) expression on PMNs at 15, 30, and 60 min postinjection of LPS were 63, 66, and 24%, respectively, compared with that on normal PMNs, indicating that the expression of TLR4 decreases in response to exposure to LPS. Our results suggest that LPS induced PMN death and that many PMNs expressing Gr-1(high) undergo apoptosis 180 min postinjection of LPS.
Collapse
Affiliation(s)
- Shuichi Miyazaki
- Department of Microbiology, Toho University School of Medicine, Omori-nishi 5-21-16, Ota-ku, Tokyo 143-8540, Japan.
| | | | | | | | | |
Collapse
|
43
|
Calder PC. n-3 fatty acids, inflammation, and immunity--relevance to postsurgical and critically ill patients. Lipids 2004; 39:1147-61. [PMID: 15736910 PMCID: PMC7101959 DOI: 10.1007/s11745-004-1342-z] [Citation(s) in RCA: 101] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2004] [Accepted: 11/26/2004] [Indexed: 01/07/2023]
Abstract
Excessive or inappropriate inflammation and immunosuppression are components of the response to surgery, trauma, injury, and infection in some individuals and these can lead, progressively, to sepsis and septic shock. The hyperinflammation is characterized by the production of inflammatory cytokines, arachidonic acid-derived eicosanoids, and other inflammatory mediators, while the immunosuppression is characterized by impairment of antigen presentation and of T helper cell type-1 responses. Long-chain n-3 FA from fish oil decrease the production of inflammatory cytokines and eicosanoids. They act both directly (by replacing arachidonic acid as an eicosanoid substrate and by inhibiting arachidonic acid metabolism) and indirectly (by altering the expression of inflammatory genes through effects on transcription factor activation). Thus, long-chain n-3 FA are potentially useful anti-inflammatory agents and may be of benefit in patients at risk of developing sepsis. As such, an emerging application of n-3 FA is in surgical or critically ill patients where they may be added to parenteral or enteral formulas. Parenteral or enteral nutrition including n-3 FA appears to preserve immune function better than standard formulas and appears to partly prevent some aspects of the inflammatory response. Studies to date are suggestive of clinical benefits from these approaches, especially in postsurgical patients.
Collapse
Affiliation(s)
- Philip C Calder
- Institute of Human Nutrition, School of Medicine, University of Southampton, Bassett Crescent East, Southampton SO16 7PX, United Kingdom.
| |
Collapse
|
44
|
Mucosal delivery of anti-inflammatory IL-1Ra by sporulating recombinant bacteria. BMC Biotechnol 2004; 4:27. [PMID: 15516267 PMCID: PMC534112 DOI: 10.1186/1472-6750-4-27] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2004] [Accepted: 10/30/2004] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Mucosal delivery of therapeutic protein drugs or vaccines is actively investigated, in order to improve bioavailability and avoid side effects associated with systemic administration. Orally administered bacteria, engineered to produce anti-inflammatory cytokines (IL-10, IL-1Ra), have shown localised ameliorating effects in inflammatory gastro-intestinal conditions. However, the possible systemic effects of mucosally delivered recombinant bacteria have not been investigated. RESULTS B. subtilis was engineered to produce the mature human IL-1 receptor antagonist (IL-1Ra). When recombinant B. subtilis was instilled in the distal colon of rats or rabbits, human IL-1Ra was found both in the intestinal lavage and in the serum of treated animals. The IL-1Ra protein in serum was intact and biologically active. IL-1-induced fever, neutrophilia, hypoglycemia and hypoferremia were inhibited in a dose-dependent fashion by intra-colon administration of IL-1Ra-producing B. subtilis. In the mouse, intra-peritoneal treatment with recombinant B. subtilis could inhibit endotoxin-induced shock and death. Instillation in the rabbit colon of another recombinant B. subtilis strain, which releases bioactive human recombinant IL-1beta upon autolysis, could induce fever and eventually death, similarly to parenteral administration of high doses of IL-1beta. CONCLUSIONS A novel system of controlled release of pharmacologically active proteins is described, which exploits bacterial autolysis in a non-permissive environment. Mucosal administration of recombinant B. subtilis causes the release of cytoplasmic recombinant proteins, which can then be found in serum and exert their biological activity in vivo systemically.
Collapse
|
45
|
Weigand MA, Hörner C, Bardenheuer HJ, Bouchon A. The systemic inflammatory response syndrome. Best Pract Res Clin Anaesthesiol 2004; 18:455-75. [PMID: 15212339 DOI: 10.1016/j.bpa.2003.12.005] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Sepsis and septic shock are the leading causes of death in intensive care units in developed countries despite recent advances in critical care medicine. Sepsis is the systemic inflammatory response to infection frequently associated with hypoperfusion followed by tissue injury and organ failure. The activation of monocytes/macrophages and neutrophils, with the consecutive release of pro-inflammatory mediators and activation of the coagulation cascade, seems to play a key role in the pathogenesis of sepsis. Elimination of the septic focus, anti-microbial therapy and supportive treatment are the cornerstones of sepsis therapy. In addition, the application of small doses hydrocortisone to patients with refractory septic shock and the treatment of patients with septic multiple organ failure with activated protein C are two adjunctive therapeutic strategies. Promising new experimental treatment options are interference with MIF, HMGB1, C5a or TREM-1 signal transduction pathways and an inhibition of apoptosis, which may further improve the prognosis of septic patients in the future.
Collapse
Affiliation(s)
- Markus A Weigand
- Department of Anesthesia, University of Heidelberg, Im Neuenheimer Feld 110, Heidelberg 69120, Germany.
| | | | | | | |
Collapse
|
46
|
Jeschke MG, Klein D, Bolder U, Einspanier R. Insulin attenuates the systemic inflammatory response in endotoxemic rats. Endocrinology 2004; 145:4084-93. [PMID: 15192048 DOI: 10.1210/en.2004-0592] [Citation(s) in RCA: 147] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Insulin decreases the mortality and prevents the incidence of infection and sepsis in critically ill patients. The molecular and cellular mechanisms by which insulin improves survival have not been defined. The purpose of the present study was to determine the effect of insulin on the inflammatory reaction during endotoxemia. Endotoxemic rats were randomly divided into two groups to receive either saline or insulin. The effects of insulin on hepatic signal transcription factor mRNA expression, proinflammatory and antiinflammatory cytokine mRNA and protein concentration were determined. Insulin administration did not change glucose or electrolyte levels, but significantly decreased proinflammatory signal transcription factors [CCAAT/enhancer-binding protein-beta, signal transducer and activator of transcription-3 and-5, RANTES (regulated on activation, normal T cell expressed and secreted)] and cytokine expression in the liver and serum levels of IL-1beta, IL-6, macrophage inflammatory factor, and TNFalpha. Insulin administration further decreased high mobility group 1 protein in the serum compared with controls. In addition, insulin increased antiinflammatory cytokine expression in the liver; serum levels of IL-2, IL-4, and IL-10; and hepatic suppressor of cytokine signaling-3 mRNA expression. Insulin modulates the inflammatory response by decreasing the proinflammatory and increasing the antiinflammatory cascade. Because glucose and electrolyte levels did not differ between insulin-treated patients and controls, we hypothesize that the effects are direct antiinflammatory mechanisms of insulin, rather than indirect, through modulation of glucose or electrolyte metabolism.
Collapse
Affiliation(s)
- Marc G Jeschke
- Department of Surgery, Friedrich Alexander University of Erlangen, Erlangen, Germany.
| | | | | | | |
Collapse
|
47
|
Iauk L, Mangano K, Rapisarda A, Ragusa S, Maiolino L, Musumeci R, Costanzo R, Serra A, Speciale A. Protection against murine endotoxemia by treatment with Ruta chalepensis L., a plant with anti-inflammatory properties. JOURNAL OF ETHNOPHARMACOLOGY 2004; 90:267-72. [PMID: 15013191 DOI: 10.1016/j.jep.2003.10.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2003] [Revised: 09/29/2003] [Accepted: 10/07/2003] [Indexed: 05/20/2023]
Abstract
The anti-inflammatory effect of the extract of Ruta Chalepensis L. (Rutaceae) on the course of lethal endotoxemia in BALB/c mice was studied. When administered by gavage as 1 g/kg per day starting 14 or 7 days prior to injection of 0.75 mg endotoxin (LPS: lipopolysaccharide), the extract markedly reduced lethality (32.5% in both experiments versus approximately 85% of the control mice). A delay in lethality, but not cumulative lethality, was observed when prophylaxis was given 24 and 1 h prior to LPS challenge. The effect was associated with reduced LPS-induced blood levels of nitrite, an indicator of nitric oxide production. In contrast, the blood levels of tumour necrosis factor, interleukin 6 and interleukin 10 did not differ significantly from those of controls given LPS alone. These data show that Ruta Chalepensis L. possesses powerful immunopharmacological properties that make it capable of counteracting the lethal effects of high doses of LPS in vivo.
Collapse
Affiliation(s)
- Liliana Iauk
- Department of Microbiological and Gynaecological Sciences, Section of Microbiology, University of Catania, Via Androne 81, Catania 95124, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Hörner C, Bouchon A, Bierhaus A, Nawroth PP, Martin E, Bardenheuer HJ, Weigand MA. [Role of the innate immune response in sepsis]. Anaesthesist 2004; 53:10-28. [PMID: 14749872 PMCID: PMC7095818 DOI: 10.1007/s00101-003-0626-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The innate immune system succeeds against the majority of infections before the adaptive immune system is activated. New findings contribute to a better understanding of the pathophysiology of sepsis and lead to the development of new therapeutic strategies. The innate immune system, being responsible for the first response to infections, can trigger adaptive immune responses in case the initial response is ineffective. Both arms of the immune system interact with each other, mainly via cell-cell-interactions but also by soluble factors, such as cytokines and chemokines. Two sub-populations of helper T-cells direct both balanced activation and inhibition of the two arms of the immune systems using specific patterns of cytokine release. Results obtained in new animal models of sepsis, taking a progressive growth of bacteria into account, have implied that existing knowledge has to be reanalyzed. The idea of sepsis as a mere "over-reaction to inflammation" has to be abandoned. Various so-called pattern recognition receptors (e.g. toll-like receptors, TLRs, NOD proteins) are located intracellularly or in the plasma membrane of innate immune cells and recognize certain patterns expressed exclusively by extracellular pathogens. Upon receptor engagement, intracellular signaling pathways lead to cellular activation, followed by release of various cytokines and anti-microbial substances. During the course of sepsis a cytokine shift towards increasing immune suppression occurs. The innate immune system also contributes to the migration of leukocytes in inflammed tissue, involving chemokines and adhesion molecules. Leukocytes also secrete the tissue factor leading to formation of thrombin. The environment in sepsis can cause disseminated intravascular coagulation (DIC), but at the same time thrombin triggers the release of chemokines and adhesion molecules through endothelial cells, which represents a positive feedback mechanism for innate immune responses. New therapeutic strategies for sepsis try to establish a well-balanced immune response. Intervention is accomplished through inhibition of inflammatory cytokines, their receptors or through activation of immunostimulatory responses.
Collapse
Affiliation(s)
- C. Hörner
- Klinik für Anaesthesiologie, Universitätsklinikum Heidelberg,
| | - A. Bouchon
- Department of Pain Research, Bayer Health Care, Wuppertal
| | - A. Bierhaus
- Abteilung Innere Medizin I, Universitätsklinikum Heidelberg,
| | - P. P. Nawroth
- Abteilung Innere Medizin I, Universitätsklinikum Heidelberg,
| | - E. Martin
- Klinik für Anaesthesiologie, Universitätsklinikum Heidelberg,
| | | | - M. A. Weigand
- Klinik für Anaesthesiologie, Universitätsklinikum Heidelberg,
- Klinik für Anaesthesiologie, Universitätsklinikum Heidelberg, Im Neuenheimer Feld 110, 69120 Heidelberg
| |
Collapse
|
49
|
Klein D, Einspanier R, Bolder U, Jeschke MG. Differences in the Hepatic Signal Transcription Pathway and Cytokine Expression Between Thermal Injury and Sepsis. Shock 2003; 20:536-43. [PMID: 14625478 DOI: 10.1097/01.shk.0000093345.68755.98] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Inflammation and catabolism in response to trauma, surgery, critical illness or bacteria lead to a compromise of essential organs, which can lead to prolonged clinical stay and even death. Mediators responsible for catabolism were thought to be proinflammatory cytokines, but recently the focus has shifted to signal transduction. The purpose of the present study was to determine differences between two pathophysiologic states, sepsis and thermal injury, in signal transduction and cytokine expression and thus define the importance of the signal transcription pathway. Rats were randomly divided to either receive lipopolysaccharide (3 mg/kg body weight or a 30% total body surface area burn) or they received no treatment and served as controls. Animals were sacrificed 1, 2, 5, and 7 days postinsult and serum and liver harvested for analysis. A thermal injury appeared to have a slow release and expression of signal transcription factors and cytokines and a sepsis showed a rapid increase of mediators and also a fast decrease. The changes in cytokine profiles after burn, particularly interleukin-1beta and macrophage inhibitory factor, appear to be mediated by C/EBP-beta and STAT-3, whereas after the induction of a sepsis, tumor necrosis factor and interleukin-6 are mainly mediated by STAT-5. Based on our findings we suggest that the pathophysiologic state of a thermal injury is not comparable with sepsis in association with signal transcription factors and the differences in intracellular and extracellular signaling therefore opens new ideas for therapeutic options.
Collapse
Affiliation(s)
- Dagmar Klein
- Klinik und Poliklinik für Chirurgie, Klinikum der Universität Regensburg, Germany
| | | | | | | |
Collapse
|
50
|
Wang JH, Doyle M, Manning BJ, Blankson S, Wu QD, Power C, Cahill R, Redmond HP. Cutting edge: bacterial lipoprotein induces endotoxin-independent tolerance to septic shock. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 170:14-8. [PMID: 12496376 DOI: 10.4049/jimmunol.170.1.14] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Tolerance to bacterial cell wall components is an adaptive host response. Endotoxin/LPS tolerance is characterized by a survival advantage against subsequent lethal LPS challenge. However, it is uncertain whether LPS tolerance can afford protection against other septic challenges. In this study, we show that tolerance induced by bacterial lipoprotein (BLP) protects mice against not only BLP-induced lethality, but also LPS-, live bacteria-, and polymicrobial sepsis-induced lethality. In contrast, LPS tolerance offers no survival benefit against the latter two challenges. Furthermore, induction of BLP tolerance results in overexpression of complement receptor type 3 and FcgammaIII/IIR on neutrophils (polymorphonuclear neutrophils) and peritoneal macrophages, with increased bacterial recognition and bactericidal activity, whereas LPS-tolerized mice exhibit an impaired ability to ingest and to kill bacteria. These results indicate that BLP tolerance is a novel adaptive host response associated with a unique protective effect during septic shock.
Collapse
Affiliation(s)
- Jiang Huai Wang
- Department of Academic Surgery, National University of Ireland, Cork University Hospital, Cork, Ireland.
| | | | | | | | | | | | | | | |
Collapse
|